Elsevier

Neurobiology of Disease

Volume 59, November 2013, Pages 183-193
Neurobiology of Disease

Pharmacological blockade of IL-1β/IL-1 receptor type 1 axis during epileptogenesis provides neuroprotection in two rat models of temporal lobe epilepsy

https://doi.org/10.1016/j.nbd.2013.07.015Get rights and content

Highlights

  • The IL-1β mediated signaling is an important target for reducing cell loss after status epilepticus.

  • Clinically tested and safe antiiflammatory drugs affords neuroprotection after a delayed post-injury intervention.

  • Early post-injury intervention is required when rapid onset inflammatory pathways are targeted.

  • Combined antiinflammatory drugs targeting different mechanisms should be considered to optimize beneficial outcomes.

Abstract

We studied whether pharmacological blockade of the IL-1β-mediated signaling, rapidly activated in forebrain by epileptogenic injuries, affords neuroprotection in two different rat models of status epilepticus (SE). As secondary outcome, we measured treatment's effect on SE-induced epileptogenesis. IL-1β signaling was blocked by systemic administration of two antiinflammatory drugs, namely human recombinant IL-1 receptor antagonist (anakinra), the naturally occurring and clinically used competitive IL-1 receptor type 1 antagonist, and VX-765 a specific non-peptide inhibitor of IL-1β cleavage and release. Antiinflammatory drugs were given 60 min after antiepileptic (AED) drug-controlled SE induced by pilocarpine, or 180 min after unrestrained electrical SE, for 7 days using a protocol yielding therapeutic drug levels in brain. This drug combination significantly decreased both IL-1β expression in astrocytes and cell loss in rat forebrain. Neuroprotection and the antiinflammatory effect were more pronounced in the electrical SE model. Onset of epilepsy, and frequency and duration of seizures 3 months after electrical SE were not significantly modified. Transcriptomic analysis in the hippocampus showed that the combined treatment did not affect the broad inflammatory response induced by SE during epileptogenesis. In particular, the treatment did not prevent the induction of the complement system and Toll-like receptors, both contributing to cell loss and seizure generation.

We conclude that the IL-1β signaling represents an important target for reducing cell loss after SE. The data highlight a new class of clinically tested agents affording neuroprotection after a delayed post-injury intervention. Earlier blockade of this rapid onset inflammatory pathway during SE, or concomitant treatment with antiinflammatory drugs targeting additional components of the broad inflammatory response to SE, or co-treatment with AEDs, is likely to be required for optimizing beneficial outcomes.

Introduction

Increased levels of inflammatory molecules and upregulation of their cognate receptors in glia, neurons and microvessels have been demonstrated in human brain specimens of drug-resistant forms of epilepsy, thus suggesting that proinflammatory pathways are activated in seizure foci (Aronica et al., 2012, Choi et al., 2009, Vezzani et al., 2011a). Induction of the same inflammatory molecules occurs after epileptogenic injuries and during recurrent seizures in epilepsy models (Aronica et al., 2012, Ravizza et al., 2011, Vezzani et al., 2013). Inhibition of experimental seizures and neuroprotection have been attained by pharmacological blockade of specific proinflammatory signalings (Aronica et al., 2012, Friedman and Dingledine, 2011, Kwon et al., 2013, Maroso et al., 2011). In particular, the activation of the IL-1 receptor 1 (IL-1R1)/Toll-like receptor (TLR) signaling in glia and neurons by the endogenous ligands IL-1β and High Mobility Group Box 1 (HMGB1) protein plays a key role in ictogenesis. In fact, treatment with the IL-1 receptor antagonist (Ra), TLR4 blockers, or specific inhibitors of Interleukin Converting Enzyme (ICE), the biosynthetic enzyme producing the releasable form of IL-1β, results in drastic reduction of acute or chronic seizures in various epilepsy models (Akin et al., 2011, Auvin et al., 2010a, Marchi et al., 2009, Maroso et al., 2010, Maroso et al., 2011, Ravizza et al., 2006, Ravizza et al., 2008b, Vezzani et al., 1999, Vezzani et al., 2000, Vezzani et al., 2002). Supportive evidence is provided by studies in transgenic mice with perturbed IL-1R1/TLR4 signals (Dubé et al., 2005, Maroso et al., 2010, Ravizza et al., 2006, Spulber et al., 2009, Vezzani et al., 2000, Vezzani et al., 2011b). There is compelling evidence that the activation of the IL-1R1/TLR4 signaling is also involved in excitotoxicity since its pharmacological blockade affords significant neuroprotection following various injuries (Allan et al., 2005, Ross et al., 2007, Vezzani et al., 2011b, Vezzani et al., 2013). In particular, IL-1β and HMGB1 enhance NMDA-induced hippocampal cell loss by increasing receptor-gated Ca2 + influx into neurons (Bernardino et al., 2008, Iori et al., 2013, Maroso et al., 2010, Viviani et al., 2003), a mechanism also involved in their proictogenic effects (Balosso et al., 2008, Maroso et al., 2010). Neuroprotective effects were observed using antagonists of the IL-1β/IL-1R1 axis in organotypic cell cultures exposed to AMPA (Bernardino et al., 2008).

The activation of the IL-1R/TLR signaling is pivotal for initiating the complex brain inflammatory response to various CNS injuries, including status epilepticus (SE) (Bartfai et al., 2007, Clausen et al., 2009, Dinarello, 2011, Ravizza et al., 2008a). This occurs by induction of NF-kB- and AP-1-dependent transcription of a large array of inflammatory genes in target cells (O'Neill and Bowie, 2007, Vezzani et al., 2011b). IL-1R1/TLR4 signaling induction after chemical or electrical SE in rodents is rapid (< 2 h) and persists until the onset of spontaneous seizures (De Simoni et al., 2000, Dhote et al., 2007, Librizzi et al., 2012, Marcon et al., 2009, Maroso et al., 2010, Ravizza et al., 2008b, Vezzani et al., 2011b, Voutsinos-Porche et al., 2004).

Beneficial outcomes, such as decreased cell loss and reduced spontaneous seizure frequency/severity, are induced by non-steroidal antiinflammatory drugs blocking pathways downstream IL-1R/TLR, when administered to rodents after SE (reviewed by Gao et al., 2012, Löscher and Brandt, 2010, Pitkanen, 2010, Ravizza et al., 2011). This evidence suggests that the various inflammatory pathways activated during epileptogenesis contribute in concert to the adverse outcomes. Preventing the activation of this inflammatory cascade by blockade of the upstream IL-1R/TLR pathway, therefore, represents a promising strategy to attain improved therapeutic effects.

In this study, we used a novel treatment combination of clinically tested and safe antiinflammatory drugs (Dinarello et al., 2012, Vezzani et al., 2010), namely human recombinant (hr)IL-1Ra (anakinra) and the ICE inhibitor VX-765, in order to block the IL-1β/IL-1R1 axis in two SE rat models. Each drug, individually, mediates neuroprotection in acute injury models (Allan et al., 2005, Ross et al., 2007), displays anti-ictogenic properties (Akin et al., 2011, Maroso et al., 2010, Maroso et al., 2011, Ravizza et al., 2006) and inhibits kindling progression (Auvin et al., 2010b, Ravizza et al., 2008b).

Our primary endpoint was to assess whether the combined treatment affords neuroprotection by preventing IL-1β actions during epileptogenesis. As a secondary outcome, we evaluated the effect of treatment on epilepsy development.

Section snippets

Animals

The experiments were carried out in two distinct laboratories: rats exposed to electrically-induced SE were prepared in Milano (male Sprague-Dawley rats, 275–300 g; Charles-River, Calco, VA, Italy) while rats exposed to lithium/pilocarpine were prepared in Hannover (female Sprague-Dawley rats, 250–275 g; Harlan, Horst, The Netherlands). Rats were housed at constant temperature (23 ± 1 °C) and relative humidity (60 ± 5%) with free access to food and water and a fixed 12 h light/dark cycle. Procedures

Plasma and CSF concentrations of anakinra after combined treatment

Fig. 1 depicts anakinra levels attained in plasma (A) and CSF (B) withdrawn from electrical SE rats undergoing combined treatment with VX-765 and anakinra for 7 days (Experiment 1). Anakinra was not detectable in SE rats treated with vehicle because the ELISA antibody recognizes only the human recombinant form, therefore it does not cross-react significantly with rat IL-1Ra. This treatment protocol induced a significant increase both in plasma and CSF anakinra levels. The attained concentrations

Discussion

This study shows that combined treatment with two clinically tested antiinflammatory drugs specifically blocking the IL-1β/IL-1R1 signaling during epileptogenesis evoked by SE, provides neuroprotection in forebrain areas although not preventing epilepsy development.

We postulated that the novel strategy of blocking the upstream IL-1β/IL-1R1 proinflammatory signaling induced by SE would prevent the activation and amplification of downstream inflammatory events involved in excitotoxicity and

Acknowledgments

The authors thank Prof. Tamas Bartfai for his review of our manuscript and suggestions. This work was supported by Nepente (Regione Lombardia, Institutional Agreement no. 14501A), Fondazione Monzino (AV) and a grant (Lo 274/11) from the Deutsche Forschungsgemeinschaft (Bonn, Germany, WL). The authors thank Dr. J. Gorter (University of Amsterdam) for his contribution in the preliminary phase of this study. We are grateful to Vertex Pharmaceuticals Inc., Cambridge, Boston, USA (VX-765) and to

References (78)

  • K.H. Jung et al.

    Cyclooxygenase-2 inhibitor, celecoxib, inhibits the altered hippocampal neurogenesis with attenuation of spontaneous recurrent seizures following pilocarpine-induced status epilepticus

    Neurobiol. Dis.

    (2006)
  • R. Kotloski et al.

    Repeated brief seizures induce progressive hippocampal neuron loss and memory deficits

    Prog. Brain Res.

    (2002)
  • M. Langer et al.

    Therapeutic window of opportunity for the neuroprotective effect of valproate versus the competitive AMPA receptor antagonist NS1209 following status epilepticus in rats

    Neuropharmacology

    (2011)
  • L. Ma et al.

    Aspirin attenuates spontaneous recurrent seizures and inhibits hippocampal neuronal loss, mossy fiber sprouting and aberrant neurogenesis following pilocarpine-induced status epilepticus in rats

    Brain Res.

    (2012)
  • N. Marchi et al.

    Antagonism of peripheral inflammation reduces the severity of status epilepticus

    Neurobiol. Dis.

    (2009)
  • J. Marcon et al.

    Age-dependent vascular changes induced by status epilepticus in rat forebrain: implications for epileptogenesis

    Neurobiol. Dis.

    (2009)
  • M. Maroso et al.

    Interleukin-1beta biosynthesis inhibition reduces acute seizures and drug resistant chronic epileptic activity in mice

    Neurotherapeutics

    (2011)
  • A. Pitkanen et al.

    Administration of diazepam during status epilepticus reduces development and severity of epilepsy in rat

    Epilepsy Res.

    (2005)
  • N. Polascheck et al.

    The COX-2 inhibitor parecoxib is neuroprotective but not antiepileptogenic in the pilocarpine model of temporal lobe epilepsy

    Exp. Neurol.

    (2010)
  • R.J. Racine

    Modification of seizure activity by electrical stimulation. II. Motor seizure

    Electroencephalogr. Clin. Neurophysiol.

    (1972)
  • T. Ravizza et al.

    Innate and adaptive immunity during epileptogenesis and spontaneous seizures: evidence from experimental models and human temporal lobe epilepsy

    Neurobiol. Dis.

    (2008)
  • T. Ravizza et al.

    Interleukin converting enzyme inhibition impairs kindling epileptogenesis in rats by blocking astrocytic IL-1beta production

    Neurobiol. Dis.

    (2008)
  • T. Ravizza et al.

    Inflammation and prevention of epileptogenesis

    Neurosci. Lett.

    (2011)
  • J. Ross et al.

    A selective, non-peptide caspase-1 inhibitor, VRT-018858, markedly reduces brain damage induced by transient ischemia in the rat

    Neuropharmacology

    (2007)
  • I. Rozovsky et al.

    Selective expression of clusterin (SGP-2) and complement C1qB and C4 during responses to neurotoxins in vivo and in vitro

    Neuroscience

    (1994)
  • C. Schmitz et al.

    Design-based stereology in neuroscience

    Neuroscience

    (2005)
  • A.K. Shetty

    Promise of resveratrol for easing status epilepticus and epilepsy

    Pharmacol. Ther.

    (2011)
  • S. Spulber et al.

    IL-1/IL-1ra balance in the brain revisited — evidence from transgenic mouse models

    Brain Behav. Immun.

    (2009)
  • A. Vezzani et al.

    IL-1 receptor/Toll-like receptor signaling in infection, inflammation, stress and neurodegeneration couples hyperexcitability and seizures

    Brain Behav. Immun.

    (2011)
  • A. Vezzani et al.

    The role of inflammation in epileptogenesis

    Neuropharmacology

    (2013)
  • B. Voutsinos-Porche et al.

    Temporal patterns of the cerebral inflammatory response in the rat lithium–pilocarpine model of temporal lobe epilepsy

    Neurobiol. Dis.

    (2004)
  • A. Alexa et al.

    Improved scoring of functional groups from gene expression data by decorrelating GO graph structure

    Bioinformatics

    (2006)
  • S.M. Allan et al.

    Interleukin-1 and neuronal injury

    Nat. Rev. Immunol.

    (2005)
  • E. Aronica et al.

    Astrocyte immune response in epilepsy

    Glia

    (2012)
  • S. Auvin et al.

    Inflammation induced by LPS enhances epileptogenesis in immature rat and may be partially reversed by IL1RA

    Epilepsia

    (2010)
  • S. Balosso et al.

    A novel non-transcriptional pathway mediates the proconvulsive effects of interleukin-1beta

    Brain

    (2008)
  • T. Bartfai et al.

    Interleukin-1 system in CNS stress: seizures, fever, and neurotrauma

    Ann. N. Y. Acad. Sci.

    (2007)
  • Y. Benjamini et al.

    Controlling the false discovery rate: a practical and powerful approach to multiple testing

    J. R. Stat. Soc.

    (1995)
  • L. Bernardino et al.

    Inflammatory events in hippocampal slice cultures prime neuronal susceptibility to excitotoxic injury: a crucial role of P2X7 receptor-mediated IL-1beta release

    J. Neurochem.

    (2008)
  • Cited by (152)

    • Microglia in epilepsy

      2023, Neurobiology of Disease
    • Neuroimmunology of status epilepticus

      2023, Epilepsy and Behavior
    • New advances in pharmacoresistant epilepsy towards precise management-from prognosis to treatments

      2022, Pharmacology and Therapeutics
      Citation Excerpt :

      The anti-seizure effect of the caspase-1 inhibitor VX765 (Wannamaker et al., 2007) has been tested. Vezzani’s group completed a series of experiments to examine the anti-seizure and anti-convulsant effect of VX765 on multiple acute and chronic animal epilepsy models (Maroso et al., 2011; Noe et al., 2013; Ravizza et al., 2006; Ravizza et al., 2008). However, it should be noted that the anti-epileptic evidences of VX765 on pharmacoresistant epilepsy models are still lacking.

    View all citing articles on Scopus
    1

    These authors contributed equally to this study.

    2

    Present address: Experimental Epileptology and Neurophysiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy.

    3

    Present address: Laboratory of Neurotoxicity and Psychopharmacology, Federal University of Santa Maria, UFSM, Santa Maria, Brazil.

    View full text